The anticancer effect and mechanism of α-hederin on breast cancer cells

  • Authors:
    • Lin Cheng
    • Tian-Song Xia
    • Yi-Fen Wang
    • Wenbin Zhou
    • Xiu-Qing Liang
    • Jin-Qiu Xue
    • Liang Shi
    • Ying Wang
    • Qiang Ding
    • Minhai Wang
  • View Affiliations

  • Published online on: May 19, 2014     https://doi.org/10.3892/ijo.2014.2449
  • Pages: 757-763
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Natural plant products occupy a very important position in the area of cancer chemotherapy. Many triterpenoid saponins have been proved as potential agents for chemoprevention and therapy of breast cancer. α-hederin, a monodesmosidic triterpenoid saponin distributed in Hedera or Nigella species, displays many biological activities. It is increasingly investigated for its promising anticancer potential since it has been shown to have cytotoxicity against several types of cancer cells. However, studies of α-hederin on breast cancer are limited, most of which focus on biological activity, while the mechanisms have not been widely reported yet. Previously, we purified and identified α-hederin from Clematis ganpiniana, a herb used in traditional Chinese medicine with antitumor action. In the present study, α-hederin showed strong inhibitory activity on the growth of breast cancer cells and induced apoptosis in these cells. α-hederin induced depolarization of mitochondrial membrane potential which released Apaf-1 and cytochrome c from the intermembrane space into the cytosol, where they promoted caspase-3 and caspase-9 activation. This is the first report on the growth inhibition and pro-apoptotic effects of α-hederin on breast cancer cells and the relative apoptosis pathways. It implied that triterpenoid saponin α-hederin could be a promising candidate for chemotherapy of breast cancer.

Introduction

Breast cancer is the most frequently diagnosed and the leading cause of cancer related death in women (1). A variety of drugs have been developed to treat breast cancer, however, drug resistance often occurs, and unexpected side-effects are common (2). Thus, novel chemotherapies that overcome drug resistance and improve patient outcomes are urgently needed. Using compounds from natural plants as potential cancer preventive and/or therapeutic agents has become a fascinating strategy (3,4). Identification and investigation of active components from natural plants are important for assessing their potential for clinical use. A large number of components purified from herbs have been used to treat various cancers including breast cancer. For example, paclitaxel (Taxol), a natural chemotherapeutic drug isolated from the bark of the pacific yew, is currently used widely for treating breast cancer (5). Therefore, development of new therapeutic agents from natural source has great promise for breast cancer treatment.

We have purified and identified four kinds of triterpenoids derivatives from Clematis ganpiniana. They showed cytotoxicity against breast cancer cells (6). One of them was α-hederin, which belonged to triterpenoid saponins. Triterpenoid saponins are an important class of natural products and distributed widely in the plant kingdom (7,8). Several excellent studies provided an overview of the triterpenoids as potential agents for chemoprevention and therapy of breast cancer (9,10).

α-hederin, a monodesmosidic triterpenoid saponin distributed in Hedera or Nigella species displays many biological activities such as anti-viral activity (11); anti-inflammatory activity (12); anti-oxidant activity (13); anti-leishmanial activity (14) and anti-spasmodic activity (15). Moreover, α-hederin is increasingly investigated for its promising anticancer potential since it revealed cytotoxicity against various cancer cell lines such as lung carcinoma, larynx epidermoid carcinoma, colon adenocarcinoma and pancreas carcinoma (1620) and in vivo tumors (2123). It has been suggested that α-hederin exerted its cytotoxic activity by promoting apoptosis and/or membrane alterations (24,25), however, the molecular and cellular mechanisms are far from being fully elucidated. Moreover, reports on the anti-breast cancer acivity of α-hederin are scarce, most of which focus on biological activity, while the mechanisms have not been widely reported yet.

α-hederin have been previously reported to inhibit growth and induce apoptosis of breast cancer cells (6), however, further effects and mechanisms of α-hederin on breast cancer is currently unavailable. In this study, we evaluated effects of α-hederin on growth and apoptosis of various human breast cancer cell lines, and explored the underlying mechanisms.

Materials and methods

Drug preparations

Protocols of the collection, storage, extraction of the plant material of Clematis ganpiniana, the methods of the purification and analysis of the α-hederin were described in a previous study (6).

Cell culture

The human breast cancer cell lines MCF-7 and MDA-MB-231 were obtained from American Type Culture Collection (Manassas, VA, USA) and incubated in a humidified atmosphere of 5% CO2 in air at 37°C and fed with the culture medium of high glucose Dulbecco’s modified Eagle’s medium, supplemented with 10% fetal bovine serum, 1% penicillin-streptomycin solution. For routine passages, cultures were split 1:3 when they reached 80–90% confluence generally every 2–3 days. All experiments were performed on exponentially growing cells. MCF-7 and MDA-MB-231 cells are widely used in studies on human breast cancer. In this study, we used these two cell lines to evaluate the growth inhibition and explore the underlying molecular mechanisms of α-hederin.

MTT assay

The MTT assay was used to measure the inhibition of growth by α-hederin in breast cancer cell lines. Briefly, 5×103 cells were seeded into a 96-well plate in triplicate and 8 h later α-hederin was added into the wells at the indicated final concentrations (0.08, 0.4, 2 and 10 μg/ml), while cells cultured in medium with 0.05% DMSO as a negative control. After incubation with α-hederin for 12, 24 and 48 h, the medium in each well was replaced with 20 μl of MTT at 5 mg/ml final concentration, and 4 h later 150 μl DMSO/well was added to dissolve the formed violet formazan crystals within metabolically viable cells. The plates were incubated at room temperature for 15 min and then read at 490 nm with a microplate reader (Tecan, Grödig, Austria). The percentage of growth inhibition was calculated as (OD of the control - OD of the experiment samples)/OD of the control × 100.

Apoptosis analysis by flow cytometry

After exposure to 2 μg/ml α-hederin for 6, 12 and 24 h, breast cancer cells were washed twice with PBS at 4°C, resuspended in stain containing Annexin V-FITC and propidium iodide (PI) for 15 min incubation on ice, and analyzed with FACSAria flow cytometer (Becton-Dickinson, San Jose, CA, USA) using FACSDiva software. Approximately 105 cells were analyzed for each treatment.

Measurement of the mitochondrial membrane potential (ΔΨm) with JC-1

The mitochondrial membrane potential was measured according to the manufacturer’s instruction with JC-1. After exposure to 2 μg/ml α-hederin for 6, 12 and 24 h, cells were washed twice with PBS, incubated in the working solution of 2 μg/ml JC-1 for 30 min at 37°C in 5% CO2 atmosphere, and observed with Zeiss LSM 5 Live confocal microscope (Carl Zeiss, Jena, Germany). The fluorescence was measured at an excitation:emission of 485/538 for green monomers and at an excitation:emission of 485/590 for red aggregates. Valinomycin was used at a concentration of 0.1 μM as a positive control for depolarization of the ΔΨm.

Measurement of cellular caspase-3 and -9 activity

Caspase-3 and -9 activity was quantified by measuring cleavage of the colorimetric peptides RED-DEVD-FMK and RED-LEHD-FMK, respectively (BioVision, Palo Alto, CA, USA). Briefly, at the end of designated treatment (24 h of exposure to 2 μg/ml α-hederin), equal number of control or treated cells were incubated with RED-DEVD-FMK and RED-LEHD-FMK, respectively (2 μg/ml) for 20 min at 37°C in 5% CO2 atmosphere, then washed twice by PBS and analyzed with FACSAria flow cytometer. For the caspase inhibition study, the cells were pre-incubated with inhibitors: z-DEVD-FMK and z-LEHD-FMK (respectively, caspase-3 and -9 inhibitors) 1 h before α-hederin treatment.

Western blot analysis

MCF-7 and MDA-MB-231 cells were seeded at 1×106 cells in 100-mm2 dishes. Cells were treated in complete medium with α-hederin for 6, 12 and 24 h. After treatment, adherent cells were gently scraped from the plates into the medium containing floating cells to obtain all the cells. Cells were then centrifuged, washed in PBS, lysed in ice-cold lysis buffer containing phosphatase inhibitor cocktail and protease inhibitor cocktail (Boehringer, Mannheim, Germany) to obtain total protein. Protein concentrations were determined using the Bradford method.

Apaf-1 and cytochrome c in mitochondrial fraction were analyzed by isolation of mitochondrial protein using the Cell Mitochondria Isolation kit (Beyotime Institute of Biotechnology, Beijing, China). Briefly, after exposure, MCF-7 and MDA-MB-231 cells were harvested and centrifuged at 800 × g at 4°C for 10 min. The pellets were added with 20 mM N-2-hydroxyethylpiperazine-N0-20-ethanesulfonic acid (HEPES) buffer containing protease inhibitor cocktail and disrupted with a glass tissue grinder. Homogenates were centrifuged at 800 × g at 4°C for 10 min, and the resulting supernatants were transferred to 0.5 ml conical tubes, and further centrifuged at 10,000 × g at 4°C for 20 min. The final pellets, containing the mitochondrial fraction, were analyzed for protein content using the Bradford method.

Cell lysates were electrophoresed through 10–12% SDS-PAGE gel, and transferred to PVDF membranes, which were activated in methanol. The blots were probed or reprobed with antibodies. GAPDH was used to normalize for protein loading. The membranes were probed using ECL and autoradiographed. The intensity of the bands was determined using densitometric analysis. The primary antibodies used were purified mouse anti-human apoptotic protease activating factor-1 (Apaf-1) and cytochrome c, purchased from BD Bioscience. β-actin was from Sigma. Anti-mouse secondary antibodies were from Cell Signaling Technology. The antibodies were diluted according to the manufacturer’s instructions.

Statistical analysis

The data were analyzed using the SPSS 20.0 software. For all the measurements, oneway ANOVA followed by Bonferroni test was used to assess the statistical significance of difference between control and groups-treated. A statistically significant difference was considered at the level of p<0.05.

Results

α-hederin inhibits the growth of breast cancer cells

In this study, two breast cancer cell lines MCF-7, MDA-MB-231, were used. The inhibitory rate of growth was determined by MTT assay. α-hederin showed inhibition in the two breast cancer cell lines which were statistically significant compared to the negative control (p<0.05) (Fig. 1).

α-hederin induces apoptosis in breast cancer cells

The apoptosis rate was measured by flow cytometry. MCF-7 and MDA-MB-231 treated with 2 μg/ml α-hederin for indicated times (6, 12 and 24 h) were first double-stained with Annexin V and PI, and then analyzed by flow cytometry. In cells treated with α-hederin, we detected a major increase in the Annexin V+/PI fraction (regarded as early apoptotic) subpopulations. After incubated with 2 μg/ml α-hederin for 24 h, early apoptosis rate of MCF-7 and MDA-MB-231 cells were significantly increased up to 25.6 and 17.0%, respectively (Fig. 2A and B). Early apoptosis rate of cells treated with α-hederin of three independent experiments are shown in column statistics (Fig. 2C).

α-hederin affects the mitochondrial membrane potential (ΔΨm) of breast cancer cells

MCF-7 and MDA-MB-231 cells were treated with 2 μg/ml α-hederin for 6, 12 and 24 h, and then mitochondrial membrane potential was measured. After the application of α-hederin, JC-1 fluorescence shifted from red-orange to greenish yellow, which indicated the depolarization of mitochondrial membrane potential (Fig. 3A). Mitochondrial membrane potential ΔΨm of cells treated with α-hederin of three independent experiments are shown in column statistics (Fig. 3B).

α-hederin regulates caspase-3 and caspase-9 activation

After exposure to α-hederin (2 μg/ml) for 24 h, activity of caspase-3 and caspase-9 was increased in both MCF-7 and MDA-MB-231 cells. This activation could be reversed by the caspase inhibitors (Fig. 4A and B). Caspase-3, and caspase-9 positive rate of cells treated with α-hederin with/without caspase inhibitors of three independent experiments are shown in column statistics (Fig. 4C and D).

α-hederin regulates the Apaf-1 and cytochrome c release

MCF-7 and MDA-MB-231 cells were treated for 2, 6, 12 or 24 h with α-hederin (2 μg/ml) and both mitochondrial Apaf-1 and cytochrome c level were detected by western blot analysis. DRβ-H decreased both mitochondrial Apaf-1 and cytochrome c expressions in a time-dependent manner (Fig. 5A). Expressions of mitochondrial Apaf-1 and cytochrome c of cells treated with α-hederin of three independent experiments are shown in column statistics (Fig. 5B and C).

Discussion

Our data indicate that α-hederin from Clematis ganpiniana had strong inhibitory activity on different breast cancer cells. α-hederin was found to induce apoptosis in both the ER+ human breast cancer cell line MCF-7 and ER breast cancer cell line MDA-MB-231. Disruption of mitochondrial membrane potential, release of Apaf-1 and cytochrome c, and subsequent activation of caspase-9 and caspase-3 was detected in α-hederin-treated cells.

The abstract of Clematis ganpiniana was traditionally used as a diuretic agent and an anti-inflammatory remedy by the Naxi people in China. α-hederin extracted from Clematis ganpiniana showed cytotoxicity on breast cancer cells. Importantly, α-hederin was found to induce apoptosis in various breast cancer cells. Apoptosis is required for proper tissue homeostasis. Defects in apoptosis signaling pathways contribute to carcinogenesis and chemoresistance. Most cancer therapeutic approaches inhibit tumors by triggering cancer cell apoptosis (26).

JC-1 staining was used to detect the membrane potential of mitochondria. The membrane potential of mitochondria in breast cancer cells was greatly reduced by α-hederin. Apaf-1 and cytochrome c were released from the mitochondria to the cytoplasm. In α-hederin-induced apoptosis, caspase-3 and caspase-9 were involved. The activation of caspase family members is a critical component of the apoptotic machinery. The caspases generally consist of the upstream initiator caspases, such as caspase-2, -8, -9 and -10, and the downstream effect of caspases, such as caspase-3, -6 and -7 (27). The results suggested that the caspase-dependent pathway mediated α-hederin-induced apoptosis in breast cancer cells through the mitochondrial pathway.

Mitochondria play a central role in cancer survival and are one of the main targets for developing anticancer drugs (28). Both the extrinsic and the intrinsic pathway can converge at the mitochondrial level and trigger mitochondrial membrane permeabilization (29). Mitochondrial apoptotic pathway was reported widely for the actions of triterpenoid saponins in other human cancers including liver cancer (3032), gastric cancer (33), esophageal cancer (34), and colorectal cancer (35). It was reported that α-hederin from Nigella sativa induced apoptosis via mitochondrial perturbations in murine leukemia P388 cells (24). We first reported mitochondrial apoptotic activity of α-hederin in breast cancer cells.

In conclusion, we showed α-hederin effectively inhibited the growth and induced apoptosis of breast cancer cells. α-hederin reduced the mitochondrial membrane potential and decreased mitochondrial Apaf-1 and cytochrome c expressions of breast cancer cells. Moreover, α-hederin increased the activity of caspase-3 and caspase-9 remarkably in breast cancer cells. Consistent with these results, α-hederin induced mitochondria-mediated apoptosis of MCF-7 and MDA-MB-231 cells. This is the first report on both chemotherapeutic effects and the mechanism of α-hederin on human breast cancer cells, which may provide a potential option for the drug development and treatment of breast cancer. Oriental medicinal herbs are rich sources of potential cancer chemopreventive and therapeutic agents. Rigorous and systematic pre-clinical evaluations in vitro was exemplified in the current study to transform traditional herbal practices into evidence-based medicine.

Acknowledgements

This study was financially supported by Natural Science Foundation of China (81272916, 81202077 and 81372828), the Natural Science Foundation of Jiangsu Province (BK2011855), the key projects of Jiangsu Provincial Health Office (H201110), the Project of Jiangsu Province Traditional Chinese Medicine Bureau (LZ11084), the Six Talents Peak projects of Jiangsu Province (to Q.D.), a project Funded by the Priority Academic Program Development of Jiangsu higher Education Institutions (PAPD) and the Talent Foundation of The First Affiliated Yijishan Hospital of Wannan Medical College (YR201305).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin. 63:11–30. 2013. View Article : Google Scholar

2 

Ribeiro JT, Macedo LT, Curigliano G, et al: Cytotoxic drugs for patients with breast cancer in the era of targeted treatment: back to the future? Ann Oncol. 23:547–555. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Karikas GA: Anticancer and chemopreventing natural products: some biochemical and therapeutic aspects. J BUON. 15:627–638. 2010.PubMed/NCBI

4 

Cragg GM and Newman DJ: Natural products: a continuing source of novel drug leads. Biochim Biophys Acta. 6:182013.

5 

Wang TH, Wang HS and Soong YK: Paclitaxel-induced cell death: Where the cell cycle and apoptosis come together. Cancer. 88:2619–2628. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Ding Q, Yang LX, Yang HW, Jiang C, Wang YF and Wang S: Cytotoxic and antibacterial triterpenoids derivatives from Clematis ganpiniana. J Ethnopharmacol. 126:382–385. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Podolak I, Galanty A and Sobolewska D: Saponins as cytotoxic agents: a review. Phytochem Rev. 9:425–474. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Augustin JM, Kuzina V, Andersen SB and Bak S: Molecular activities, biosynthesis and evolution of triterpenoid saponins. Phytochemistry. 72:435–457. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Bishayee A, Ahmed S, Brankov N and Perloff M: Triterpenoids as potential agents for the chemoprevention and therapy of breast cancer. Front Biosci (Landmark Ed). 16:980–996. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Patlolla JMR and Rao CV: Triterpenoids for cancer prevention and treatment: Current status and future prospects. Curr Pharm Biotechnol. 13:147–155. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Calabrese AI: Letter: Antiviral activity of hederin. J Pharm Sci. 64:VIII1975. View Article : Google Scholar : PubMed/NCBI

12 

Gepdiremen A, Mshvildadze V, Suleyman H and Elias R: Acute anti-inflammatory activity of four saponins isolated from ivy: alpha-hederin, hederasaponin-C, hederacolchiside-E and hederacolchiside-F in carrageenan-induced rat paw edema. Phytomedicine. 12:440–444. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Gulcin I, Mshvildadze V, Gepdiremen A and Elias R: Antioxidant activity of saponins isolated from ivy: alpha-hederin, hederasaponin-C, hederacolchiside-E and hederacolchiside-F. Planta Med. 70:561–563. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Ridoux O, Di Giorgio C, Delmas F, et al: In vitro antileishmanial activity of three saponins isolated from ivy, alpha-hederin, beta-hederin and hederacolchiside A(1), in association with pentamidine and amphotericin B. Phytother Res. 15:298–301. 2001. View Article : Google Scholar : PubMed/NCBI

15 

Trute A, Gross J, Mutschler E and Nahrstedt A: In vitro antispasmodic compounds of the dry extract obtained from Hedera helix. Planta Med. 63:125–129. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Quetin-Leclercq J, Elias R, Balansard G, Bassleer R and Angenot L: Cytotoxic activity of some triterpenoid saponins. Planta Med. 58:279–281. 1992. View Article : Google Scholar : PubMed/NCBI

17 

Danloy S, Quetin-Leclercq J, Coucke P, et al: Effects of alpha-hederin, a saponin extracted from Hedera helix, on cells cultured in vitro. Planta Med. 60:45–49. 1994. View Article : Google Scholar

18 

Rooney S and Ryan MF: Effects of alpha-hederin and thymoquinone, constituents of Nigella sativa, on human cancer cell lines. Anticancer Res. 25:2199–2204. 2005.PubMed/NCBI

19 

Tian Z, Liu YM, Chen SB, et al: Cytotoxicity of two triterpenoids from Nigella glandulifera. Molecules. 11:693–699. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Yan LH, Xu LZ, Lin J, Yang SL and Feng YL: Triterpenoid saponins from the stems of Clematis parviloba. J Asian Nat Prod Res. 11:332–338. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Kumara SS and Huat BT: Extraction, isolation and characterisation of antitumor principle, alpha-hederin, from the seeds of Nigella sativa. Planta Med. 67:29–32. 2001. View Article : Google Scholar : PubMed/NCBI

22 

Feller G, Kugel A, Moonshine D, et al: African descents are more sensitive than European descents to the antitumor compounds alpha-hederin and kalopanaxsaponin I. Planta Med. 76:1847–1851. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Pasi S, Aligiannis N, Pratsinis H, Skaltsounis AL and Chinou IB: Biologically active triterpenoids from Cephalaria ambrosioides. Planta Med. 75:163–167. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Swamy SM and Huat BT: Intracellular glutathione depletion and reactive oxygen species generation are important in alpha-hederin-induced apoptosis of P388 cells. Mol Cell Biochem. 245:127–139. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Rooney S and Ryan MF: Modes of action of alpha-hederin and thymoquinone, active constituents of Nigella sativa, against HEp-2 cancer cells. Anticancer Res. 25:4255–4259. 2005.PubMed/NCBI

26 

Liu JJ, Lin M, Yu JY, Liu B and Bao JK: Targeting apoptotic and autophagic pathways for cancer therapeutics. Cancer Lett. 300:105–114. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Shi Y: Mechanisms of caspase activation and inhibition during apoptosis. Mol Cell. 9:459–470. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Dias N and Bailly C: Drugs targeting mitochondrial functions to control tumor cell growth. Biochem Pharmacol. 70:1–12. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Suen DF, Norris KL and Youle RJ: Mitochondrial dynamics and apoptosis. Genes Dev. 22:1577–1590. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Wang QF, Chen JC, Hsieh SJ, Cheng CC and Hsu SL: Regulation of Bcl-2 family molecules and activation of caspase cascade involved in gypenosides-induced apoptosis in human hepatoma cells. Cancer Lett. 183:169–178. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Wang J, Zhao XZ, Qi Q, et al: Macranthoside B, a hederagenin saponin extracted from Lonicera macranthoides and its anti-tumor activities in vitro and in vivo. Food Chem Toxicol. 47:1716–1721. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Park HM, Kim SJ, Kim JS and Kang HS: Reactive oxygen species mediated ginsenoside Rg3- and Rh2-induced apoptosis in hepatoma cells through mitochondrial signaling pathways. Food Chem Toxicol. 50:2736–2741. 2012. View Article : Google Scholar

33 

Chun J, Ha IJ and Kim YS: Antiproliferative and apoptotic activities of triterpenoid saponins from the roots of Platycodon grandiflorum and their structure-activity relationships. Planta Med. 79:639–645. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Mo S, Xiong H, Shu G, et al: Phaseoloideside E, a novel natural triterpenoid saponin identified from Entada phaseoloides, induces apoptosis in Ec-109 esophageal cancer cells through reactive oxygen species generation. J Pharmacol Sci. 122:163–175. 2013.PubMed/NCBI

35 

Wang CZ, Li XL, Wang QF, Mehendale SR, Fishbein AB, Han AH, Sun S and Yuan CS: The mitochondrial pathway is involved in American ginseng-induced apoptosis of SW-480 colon cancer cells. Oncol Rep. 21:577–584. 2009.PubMed/NCBI

Related Articles

Journal Cover

August-2014
Volume 45 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cheng L, Xia T, Wang Y, Zhou W, Liang X, Xue J, Shi L, Wang Y, Ding Q, Wang M, Wang M, et al: The anticancer effect and mechanism of α-hederin on breast cancer cells. Int J Oncol 45: 757-763, 2014
APA
Cheng, L., Xia, T., Wang, Y., Zhou, W., Liang, X., Xue, J. ... Wang, M. (2014). The anticancer effect and mechanism of α-hederin on breast cancer cells. International Journal of Oncology, 45, 757-763. https://doi.org/10.3892/ijo.2014.2449
MLA
Cheng, L., Xia, T., Wang, Y., Zhou, W., Liang, X., Xue, J., Shi, L., Wang, Y., Ding, Q., Wang, M."The anticancer effect and mechanism of α-hederin on breast cancer cells". International Journal of Oncology 45.2 (2014): 757-763.
Chicago
Cheng, L., Xia, T., Wang, Y., Zhou, W., Liang, X., Xue, J., Shi, L., Wang, Y., Ding, Q., Wang, M."The anticancer effect and mechanism of α-hederin on breast cancer cells". International Journal of Oncology 45, no. 2 (2014): 757-763. https://doi.org/10.3892/ijo.2014.2449